Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Oncol ; 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38978350

ABSTRACT

Paired related homeobox 1 (PRRX1) is an inducer of epithelial-to-mesenchymal transition (EMT) in different types of cancer cells. We detected low PRRX1 expression in nevus but increased levels in primary human melanoma and cell lines carrying the BRAFV600E mutation. High expression of PRRX1 correlates with invasiveness and enrichment of genes belonging to the EMT programme. Conversely, we found that loss of PRRX1 in metastatic samples is an independent prognostic predictor of poor survival for melanoma patients. Here, we show that stable depletion of PRRX1 improves the growth of melanoma xenografts and increases the number of distant spontaneous metastases, compared to controls. We provide evidence that loss of PRRX1 counteracts the EMT phenotype, impairing the expression of other EMT-related transcription factors, causing dysregulation of the ERK and signal transducer and activator of transcription 3 (STAT3) signaling pathways, and abrogating the invasive and migratory properties of melanoma cells while triggering the up-regulation of proliferative/melanocytic genes and the expression of the neural-crest-like markers nerve growth factor receptor (NGFR; also known as neurotrophin receptor p75NTR) and neural cell adhesion molecule L1 (L1CAM). Overall, our results indicate that loss of PRRX1 triggers a switch in the invasive programme, and cells de-differentiate towards a neural crest stem cell (NCSC)-like phenotype that accounts for the metastatic aggressiveness.

2.
Melanoma Res ; 33(1): 1-11, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36302215

ABSTRACT

Uveal melanoma is the most common intraocular malignancy in adults. Despite the effective primary treatment, up to 50% of patients with uveal melanoma will develop metastatic lesions mainly in the liver, which are resistant to conventional chemotherapy and lead to patient's death. To date, no orthotopic murine models of uveal melanoma which can develop spontaneous metastasis are available for preclinical studies. Here, we describe a spontaneous metastatic model of uveal melanoma based on the orthotopic injection of human uveal melanoma cells into the suprachoroidal space of immunodeficient NSG mice. All mice injected with bioluminescent OMM2.5 ( n = 23) or MP41 ( n = 19) cells developed a primary tumor. After eye enucleation, additional bioluminescence signals were detected in the lungs and in the liver. At necropsy, histopathological studies confirmed the presence of lung metastases in 100% of the mice. Liver metastases were assessed in 87 and in 100% of the mice that received OMM2.5 or MP41 cells, respectively. All tumors and metastatic lesions expressed melanoma markers and the signaling molecules insulin-like growth factor type I receptor and myristoylated alanine-rich C-kinase substrate, commonly activated in uveal melanoma. The novelty of this orthotopic mouse xenograft model is the development of spontaneous metastases in the liver from the primary site, reproducing the organoespecificity of metastasis observed in uveal melanoma patients. The faster growth and the high metastatic incidence may be attributed at least in part, to the severe immunodeficiency of NSG mice. This model may be useful for preclinical testing of targeted therapies with potential uveal melanoma antimetastatic activity and to study the mechanisms involved in liver metastasis.


Subject(s)
Liver Neoplasms , Melanoma , Skin Neoplasms , Uveal Neoplasms , Adult , Humans , Animals , Mice , Melanoma/pathology , Heterografts , Disease Models, Animal , Uveal Neoplasms/pathology , Liver Neoplasms/secondary
3.
Pigment Cell Melanoma Res ; 35(4): 450-460, 2022 07.
Article in English | MEDLINE | ID: mdl-35587097

ABSTRACT

We recently reported an RAF rearrangement without NRAS or BRAF mutations in lesions from Giant Congenital Melanocytic Nevi (CMN). The new gene fusion involves the 5'-end of the promoter-containing N terminus of the SOX5 gene fused to exons 7-16 of the 3'-end of RAF1 gene leading to a SOX5-RAF1 fusion transcript which loses the auto-inhibitory CR1 domain but retains the complete in-frame coding sequence for the C-Terminal kinase domain of the RAF1. Stable expression of SOX5-RAF1 fusion induced growth factor-independent cell growth in murine hematopoietic Ba/F3 cells and melan-a immortalized melanocytes. Besides, it led to the transformation of both Ba/F3 and NIH 3T3 cells as revealed by colony formation assays. Furthermore, its expression results in MAPK activation assessed by increased levels of p-ERK protein in the cytosol of transduced cells. Treatment with Sorafenib and UO126 inhibited proliferation of Ba/F3-SOX5-RAF1 cells in the absence of IL3 but not the PLX 4720, a specific inhibitor of BRAF. Moreover, the tumorigenic and metastatic capacities of SOX5-RAF1 were assessed in vivo. These results indicate that SOX5-RAF1, a driver event for CMN development, has oncogenic capacity. Thus, sequencing of CMN transcriptomes may lead to the identification of this druggable fusion and interfere with the progression toward melanoma.


Subject(s)
MAP Kinase Signaling System , Nevus, Pigmented , Proto-Oncogene Proteins c-raf/genetics , SOXD Transcription Factors/genetics , Skin Neoplasms , Animals , Gene Fusion , Mice , Monomeric GTP-Binding Proteins/metabolism , Mutation , Nevus, Pigmented/pathology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/pathology
4.
Cancers (Basel) ; 14(5)2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35267510

ABSTRACT

Malignant melanoma is a highly aggressive tumor causing most skin cancer-related deaths. Understanding the fundamental mechanisms responsible for melanoma progression and therapeutic evasion is still an unmet need for melanoma patients. Progression of skin melanoma and its dissemination to local or distant organs relies on phenotypic plasticity of melanoma cells, orchestrated by EMT-TFs and microphthalmia-associated TF (MITF). Recently, melanoma phenotypic switching has been proposed to uphold context-dependent intermediate cell states benefitting malignancy. LOXL3 (lysyl oxidase-like 3) promotes EMT and has a key role in human melanoma cell survival and maintenance of genomic integrity. To further understand the role of Loxl3 in melanoma, we generated a conditional Loxl3-knockout (KO) melanoma mouse model in the context of BrafV600E-activating mutation and Pten loss. Melanocyte-Loxl3 deletion increased melanoma latency, decreased tumor growth, and reduced lymph node metastatic dissemination. Complementary in vitro and in vivo studies in mouse melanoma cells confirmed Loxl3's contribution to melanoma progression and metastasis, in part by modulating phenotypic switching through Snail1 and Prrx1 EMT-TFs. Importantly, a novel LOXL3-SNAIL1-PRRX1 axis was identified in human melanoma, plausibly relevant to melanoma cellular plasticity. These data reinforced the value of LOXL3 as a therapeutic target in melanoma.

5.
J Invest Dermatol ; 137(6): 1297-1310, 2017 06.
Article in English | MEDLINE | ID: mdl-28188776

ABSTRACT

The cell cycle-related genes AURKA and FOXM1 are overexpressed in melanoma. We show here that AURKA overexpression is associated with poor prognosis in three independent cohorts of melanoma patients and correlates with the presence of genomic amplification of AURKA locus and BRAFV600E mutation. AURKA overexpression may also be driven by increased promoter activation through elements such as ETS and FOXM1 found within the 5' proximal promoter region. Activated MAPK/ERK signaling pathway mediates robust AURKA promoter activation, thereby knockdown of BRAFV600E and ERK inhibition results in reduced AURKA transcription and expression. We show a positive correlation between FOXM1 and AURKA expression in three independent cohorts of melanoma patients. FOXM1 silencing decreases expression of AURKA and late cell cycle genes in melanoma cells. We further found that FOXM1 expression levels are significantly higher in tumors carrying the BRAFV600E mutation compared with the wild-type BRAF (BRAFwt). Accordingly, the knockdown of BRAFV600E also reduces the expression of FOXM1 in BRAFV600E cells. Moreover, Aurora kinase A and FOXM1 inhibition by either genetic knockdown or pharmacologic inhibitors impair melanoma growth and survival both in culture and in vivo, underscoring their therapeutic value for melanoma patients who fail to benefit from BRAF/MEK signaling inhibition.


Subject(s)
Aspartate-tRNA Ligase/genetics , Aurora Kinase A/genetics , Forkhead Box Protein M1/metabolism , Gene Expression Regulation, Neoplastic , Mutation , RNA, Transfer, Amino Acyl/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Humans , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins B-raf/genetics , Sensitivity and Specificity , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/therapy , Xenograft Model Antitumor Assays
6.
Cell Signal ; 21(11): 1595-606, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19586611

ABSTRACT

Treatment of FaO rat hepatoma cells with TGF-beta selects cells that survive to its apoptotic effect and undergo epithelial-mesenchymal transitions (EMT). We have established a cell line (T beta T-FaO, from TGF-beta-treated FaO) that shows a mesenchymal, de-differentiated, phenotype in the presence of TGF-beta and is refractory to its suppressor effects. In the absence of this cytokine, cells revert to an epithelial phenotype in 3-4 weeks and recover the response to TGF-beta. T beta T-FaO show higher capacity to migrate than that observed in the parental FaO cells. We found that FaO cells express low levels of CXCR4 and do not respond to SDF-1 alpha. However, TGF-beta up-regulates CXCR4, through a NF kappaB-dependent mechanism, and T beta T-FaO cells show elevated levels of CXCR4, which is located in the presumptive migration front. A specific CXCR4 antagonist (AMD3100) attenuates the migratory capacity of T beta T-FaO cells on collagen gels. Extracellular SDF-1 alpha activates the ERKs pathway in T beta T-FaO, but not in FaO cells, increasing cell scattering and protecting cells from apoptosis induced by serum deprivation. Targeted knock-down of CXCR4 with specific siRNA blocks the T beta T-FaO response to SDF-1 alpha. Thus, the SDF-1/CXCR4 axis might play an important role in mediating cell migration and survival after a TGF-beta-induced EMT in hepatoma cells.


Subject(s)
Chemokine CXCL12/physiology , Liver Neoplasms, Experimental/metabolism , Receptors, CXCR4/physiology , Transforming Growth Factor beta/pharmacology , Animals , Apoptosis , Benzylamines , Cell Differentiation , Cell Line, Tumor , Cell Movement , Chemokine CXCL12/metabolism , Cyclams , Epithelial Cells/drug effects , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Heterocyclic Compounds/pharmacology , Liver Neoplasms, Experimental/pathology , Mesoderm/pathology , NF-kappa B/metabolism , Phenotype , RNA, Small Interfering/metabolism , Rats , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction
7.
Exp Cell Res ; 313(11): 2389-403, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17490644

ABSTRACT

Id-1, a member of the helix-loop-helix transcription factor family has been shown to be involved in cell proliferation, angiogenesis and invasion of many types of human cancers. We have previously shown that stable expression of E47 and Snail repressors of the E-cadherin promoter in MDCK epithelial cell line triggers epithelial mesenchymal transition (EMT) concomitantly with changes in gene expression. We show here that both factors activate the Id-1 gene promoter and induce Id-1 mRNA and protein. The upregulation of the Id-1 gene occurs through the transactivation of the promoter by the Erk/MAPK signaling pathway. Moreover, oncogenic Ras is also able to activate Id-1 promoter in MDCK cells in the absence of both E47 and Snail transcription factors. Several transcriptionally active regulatory elements have been identified in the proximal promoter, including AP-1, Sp1 and four putative E-boxes. By EMSA, we only detected an increased binding to Sp1 and AP-1 elements in E47- and Snail-expressing cells. Binding is affected by the treatment of cells with PD 98059 MEK inhibitor, suggesting that MAPK/Erk contributes to the recruitment or assembly of proteins to Id-1 promoter. Small interfering RNA directed against Sp1 reduced Id-1 expression and the upregulation of the promoter, indicating that Sp1 is required for Id-1 induction in E47- and Snail-expressing cells. Our results provide new insights into how some target genes are activated during and/or as a consequence of the EMT triggered by both E47 and Snail transcription factors.


Subject(s)
Gene Expression Regulation , Inhibitor of Differentiation Protein 1/genetics , TCF Transcription Factors/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Dogs , Electrophoretic Mobility Shift Assay , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Inhibitor of Differentiation Protein 1/biosynthesis , MAP Kinase Signaling System , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Snail Family Transcription Factors , Sp1 Transcription Factor/metabolism , TCF Transcription Factors/genetics , Transcription Factor 7-Like 1 Protein , Transcription Factor AP-1/metabolism , Transcription Factors/genetics
8.
Int J Biochem Cell Biol ; 38(4): 544-62, 2006.
Article in English | MEDLINE | ID: mdl-16343978

ABSTRACT

We analyzed the differential gene expression between variants of MDA-MB-435 human breast cancer cell line that share an identical genetic background but have different metastatic ability. The major histocompatibility complex class II was found down-regulated in highly metastatic cells and correlated with MHC transactivator (CIITA) expression. Constitutive CIITA expression observed in poorly metastatic is driven by promoters III and IV of CIITA gene. Conversely, both promoters were ineffective in highly metastatic cells. The MHC class II and CIITA expression was restored in these cells upon stimulation with IFNgamma or by the treatment with a hypomethylating agent. Both treatments induced USF-1 and IRF binding complexes to promoter IV but only IFNgamma induced the binding of 435-Lung2 nuclear proteins to an ARE-1 site at the promoter III. Neither Southern blot nor bisulfite sequencing of promoter IV demonstrated strong hypermethylation of this promoter at the IFNgamma-responsive elements such as GAS, E-box or IRF-1. We suggest that partial or hemimethylation of promoter IV is sufficient to silence the CIITA expression in highly metastatic cells and that this epigenetic mechanism is responsible for the lack of MHC-II expression. Forced CIITA expression restored the MHC-II antigen expression in 435-Lung2 cells and abrogates spontaneous lung metastasis in both SCID and nude mice but also affected the tumorigenicity in nude mice. The increase in NK cell infiltration in nude mice bearing CIITA-tumors correlated with sign of tumor cell apoptosis and the increase in the number of NK cells in the spleens, suggesting that NK cells might be responsible for the observed antitumor activity.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class II/biosynthesis , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , Apoptosis/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , DNA Methylation , Female , Histocompatibility Antigens Class II/immunology , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Mice , Mice, Nude , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasm Proteins/immunology , Neoplasm Transplantation , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nuclear Proteins/immunology , Response Elements/immunology , Trans-Activators/immunology
9.
J Cell Sci ; 118(Pt 15): 3371-85, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16079281

ABSTRACT

Overexpression of the transcription factor Snail in epithelial MDCK cells promotes the epithelial-mesenchymal transition (EMT) and the acquisition of an invasive phenotype. We report here that the expression of Snail is associated with an increase in the promoter activity and expression of the matrix metalloproteinase MMP-9. The effect of Snail silencing on MMP-9 expression corroborates this finding. Induced transcription of MMP-9 by Snail is driven by a mechanism dependent on the MAPK and phosphoinositide 3-kinase (PI3K) signalling pathways. Although other regions of the promoter were required for a complete stimulation by Snail, a minimal fragment (nucleotides -97 to +114) produces a response following an increased phosphorylation of Sp-1 and either Sp-1 or Ets-1 binding to the GC-box elements contained in this region. The expression of a dominant negative form of MEK decreased these complexes. A moderate increase in the binding of the nuclear factor kappaB (NFkappaB) to the upstream region (nucleotide -562) of the MMP-9 promoter was also observed in Snail-expressing cells. Interestingly, oncogenic H-Ras (RasV12) synergistically co-operates with Snail in the induction of MMP-9 transcription and expression. Altogether, these results indicate that MMP-9 transcription is activated in response to Snail expression and that it might explain, at least in part, the invasive properties of the Snail-expressing cells.


Subject(s)
Epithelial Cells/metabolism , Kidney Tubules/physiology , Matrix Metalloproteinase 9/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation/physiology , Animals , Cell Line , Dogs , Epithelial Cells/cytology , Gene Silencing , Genes, ras/physiology , Kidney Tubules/cytology , Matrix Metalloproteinase 9/drug effects , Protein Binding , Signal Transduction/drug effects , Signal Transduction/physiology , Snail Family Transcription Factors , Sp1 Transcription Factor/metabolism , Transcription Factors/pharmacology , Transcription Factors/physiology , Up-Regulation/genetics
10.
J Invest Dermatol ; 123(6): 1151-61, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15610528

ABSTRACT

Vascular endothelial growth factor (VEGF) is an important mediator of tumor-associated angiogenesis, and consequently it has been associated with metastasis. We report here that the overexpression of VEGF(165) in melanoma xenografts promotes an acceleration of tumor growth and an increase in angiogenesis as well as the spontaneous metastasis formation. In addition, VEGF receptors (VEGFR)1, VEGFR2 and neurophilin-1 are expressed in A375 melanoma cells. Forced overexpression of VEGF in these cells induces cell growth and triggers survival activity in serum-starved cultures, by a mechanism dependent on the mitogen-activating protein kinase signaling pathway. Furthermore, these effects are dependent MEK 1/2 activity. Kinase domain region-specific tyrosine kinase inhibitors dramatically reduced DNA synthesis to 20% with respect to the controls, although they did not completely suppress either the p44 or p42-phosphorylated forms of extracellular signal-regulated protein kinase. These inhibitors also provoked a decrease in Akt phosphorylation. We observed a dramatic reduction in survival after treatment with phosphatidylinositol 3'-kinase (PI3K)-specific inhibitor in the presence of specific tyrosinase inhibitors. We suggest that the overproduction of VEGF(165) concomitantly expressed with its receptors favors cell growth and survival of melanoma cells through MAPK and PI3K signaling pathways. These data support the involvement in melanoma growth and survival of a VEGF-dependent internal autocrine loop mechanism, at least in vitro.


Subject(s)
MAP Kinase Signaling System/physiology , Melanoma , Skin Neoplasms , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Cell Division/physiology , Cell Line, Tumor , Cell Survival/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Kinetics , Neovascularization, Pathologic/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...